Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where C. Ryan Miller is active.

Publication


Featured researches published by C. Ryan Miller.


Cancer Cell | 2010

Integrated Genomic Analysis Identifies Clinically Relevant Subtypes of Glioblastoma Characterized by Abnormalities in PDGFRA, IDH1, EGFR, and NF1

Roel G.W. Verhaak; Katherine A. Hoadley; Elizabeth Purdom; Victoria Wang; Yuan Qi; Matthew D. Wilkerson; C. Ryan Miller; Li Ding; Todd R. Golub; Jill P. Mesirov; Gabriele Alexe; Michael S. Lawrence; Michael O'Kelly; Pablo Tamayo; Barbara A. Weir; Stacey Gabriel; Wendy Winckler; Supriya Gupta; Lakshmi Jakkula; Heidi S. Feiler; J. Graeme Hodgson; C. David James; Jann N. Sarkaria; Cameron Brennan; Ari Kahn; Paul T. Spellman; Richard Wilson; Terence P. Speed; Joe W. Gray; Matthew Meyerson

The Cancer Genome Atlas Network recently cataloged recurrent genomic abnormalities in glioblastoma multiforme (GBM). We describe a robust gene expression-based molecular classification of GBM into Proneural, Neural, Classical, and Mesenchymal subtypes and integrate multidimensional genomic data to establish patterns of somatic mutations and DNA copy number. Aberrations and gene expression of EGFR, NF1, and PDGFRA/IDH1 each define the Classical, Mesenchymal, and Proneural subtypes, respectively. Gene signatures of normal brain cell types show a strong relationship between subtypes and different neural lineages. Additionally, response to aggressive therapy differs by subtype, with the greatest benefit in the Classical subtype and no benefit in the Proneural subtype. We provide a framework that unifies transcriptomic and genomic dimensions for GBM molecular stratification with important implications for future studies.


Cancer Cell | 2012

An Animal Model of MYC-Driven Medulloblastoma

Yanxin Pei; Colin Moore; Jun Wang; Alok K. Tewari; Alexey Eroshkin; Yoon-Jae Cho; Hendrik Witt; Andrey Korshunov; Tracy Ann Read; Julia L. Sun; Earlene M. Schmitt; C. Ryan Miller; Anne F. Buckley; Roger E. McLendon; Thomas F. Westbrook; Paul A. Northcott; Michael D. Taylor; Stefan M. Pfister; Phillip G. Febbo; Robert J. Wechsler-Reya

Medulloblastoma (MB) is the most common malignant brain tumor in children. Patients whose tumors exhibit overexpression or amplification of the MYC oncogene (c-MYC) usually have an extremely poor prognosis, but there are no animal models of this subtype of the disease. Here, we show that cerebellar stem cells expressing Myc and mutant Trp53 (p53) generate aggressive tumors following orthotopic transplantation. These tumors consist of large, pleiomorphic cells and resemble human MYC-driven MB at a molecular level. Notably, antagonists of PI3K/mTOR signaling, but not Hedgehog signaling, inhibit growth of tumor cells. These findings suggest that cerebellar stem cells can give rise to MYC-driven MB and identify a novel model that can be used to test therapies for this devastating disease.


Journal of Clinical Oncology | 2006

Significance of Necrosis in Grading of Oligodendroglial Neoplasms: A Clinicopathologic and Genetic Study of Newly Diagnosed High-Grade Gliomas

C. Ryan Miller; Christopher Dunham; Bernd W. Scheithauer; Arie Perry

PURPOSE High-grade gliomas (HGGs; WHO grades 3-4) are highly diverse, with survival times ranging from months to years. WHO 2000 grading criteria for high-grade oligodendroglial neoplasms [anaplastic oligoastrocytoma (AOA) and anaplastic oligodendroglioma (AO)] remain subjective, and the existence of grade 4 variants is controversial. PATIENTS AND METHODS Overall survival (OS) of 1,093 adult patients with a cerebral HGG newly diagnosed between 1990 and 2005 was analyzed by univariate and multivariate models for significance of the following factors: patient age, surgery type, year of diagnosis, endothelial proliferation, necrosis, oligodendroglial histology, treatment center, and chromosome 1p, 19q, 7p (EGFR), and 10q (PTEN) abnormalities by fluorescence in situ hybridization (FISH). RESULTS Necrosis was a statistically significant predictor of poor OS on univariate and multivariate analyses in AOA but not in AO. Median OS for patients with necrotic AOA (22.8 months) was significantly worse than for patients with non-necrotic AOA (86.9 months; P < .0001) but was better than conventional glioblastomas (9.8 months; P < .0001). In addition to patient age, the following were significant independent prognostic factors (P .001): grade and surgery type for the entire HGG cohort; modified grade for AOA (3 v 4); and modified grade, 1p/19q codeletion status, and oligodendroglial histology for the 586 HGGs analyzed by FISH. CONCLUSION Stratification of AOA, but not of pure AO, into grades 3 and 4 on the basis of necrosis is prognostically justified and is more powerful than the current approach. Both routine histology and genetic testing provide independent, prognostically useful information.


Nature Biotechnology | 1999

A system for the propagation of adenoviral vectors with genetically modified receptor specificities

Joanne T. Douglas; C. Ryan Miller; Myunghee Kim; Igor Dmitriev; Galina Mikheeva; Victor Krasnykh; David T. Curiel

The development of genetically modified adenovirus (Ad) vectors with specificity for a single cell type will require both the introduction of novel tropism determinants and the ablation of endogenous tropism. Consequently, it will not be possible to exploit the native cellular entry pathway in the propagation of these targeted Ad vectors. Based on the concept that Ad enters cells by a two-step process in which a primary receptor serves as a high affinity binding site for the Ad fiber knob, with subsequent internalization mediated by αv integrins, we designed two artificial primary receptors. The extracellular domain of one of these synthetic receptors was derived from a single-chain antibody (sFv) with specificity for Ad5 knob, while the second receptor consisted of an icosapeptide identified by biopanning a phage display library against Ad5 knob. Expression of either of these artificial virus-binding receptors in fiber receptor-negative cells possessing αv integrins conferred susceptibility to Ad infection. We then created a novel mechanism for cell binding by genetically modifying both the vector and the target cell. In this approach, six histidine (His) residues were incorporated at the C-terminal of the Ad fiber protein. The resultant Ad vector was able to infect nonpermissive cells displaying the cognate artificial receptor, containing an anti-His sFv. This strategy, comprising a genetically engineered Ad virion and a modified cell line, should be useful in the propagation of targeted Ad vectors that lack the ability to bind the native fiber receptor.


Clinical Cancer Research | 2010

Lung squamous cell carcinoma mRNA expression subtypes are reproducible, clinically important, and correspond to normal cell types.

Matthew D. Wilkerson; Xiaoying Yin; Katherine A. Hoadley; Yufeng Liu; Michele C. Hayward; Christopher R. Cabanski; Kenneth L. Muldrew; C. Ryan Miller; Scott H. Randell; Mark A. Socinski; Alden M. Parsons; William K. Funkhouser; Carrie B. Lee; Patrick J. Roberts; Leigh B. Thorne; Philip S. Bernard; Charles M. Perou; D. Neil Hayes

Purpose: Lung squamous cell carcinoma (SCC) is clinically and genetically heterogeneous, and current diagnostic practices do not adequately substratify this heterogeneity. A robust, biologically based SCC subclassification may describe this variability and lead to more precise patient prognosis and management. We sought to determine if SCC mRNA expression subtypes exist, are reproducible across multiple patient cohorts, and are clinically relevant. Experimental Design: Subtypes were detected by unsupervised consensus clustering in five published discovery cohorts of mRNA microarrays, totaling 382 SCC patients. An independent validation cohort of 56 SCC patients was collected and assayed by microarrays. A nearest-centroid subtype predictor was built using discovery cohorts. Validation cohort subtypes were predicted and evaluated for confirmation. Subtype survival outcome, clinical covariates, and biological processes were compared by statistical and bioinformatic methods. Results: Four lung SCC mRNA expression subtypes, named primitive, classical, secretory, and basal, were detected and independently validated (P < 0.001). The primitive subtype had the worst survival outcome (P < 0.05) and is an independent predictor of survival (P < 0.05). Tumor differentiation and patient sex were associated with subtype. The expression profiles of the subtypes contained distinct biological processes (primitive: proliferation; classical: xenobiotic metabolism; secretory: immune response; basal: cell adhesion) and suggested distinct pharmacologic interventions. Comparison with lung model systems revealed distinct subtype to cell type correspondence. Conclusions: Lung SCC consists of four mRNA expression subtypes that have different survival outcomes, patient populations, and biological processes. The subtypes stratify patients for more precise prognosis and targeted research. Clin Cancer Res; 16(19); 4864–75. ©2010 AACR.


PLOS ONE | 2012

Differential Pathogenesis of Lung Adenocarcinoma Subtypes Involving Sequence Mutations, Copy Number, Chromosomal Instability, and Methylation

Matthew D. Wilkerson; Xiaoying Yin; Vonn Walter; Ni Zhao; Christopher R. Cabanski; Michele C. Hayward; C. Ryan Miller; Mark A. Socinski; Alden M. Parsons; Leigh B. Thorne; Benjamin E. Haithcock; Nirmal K. Veeramachaneni; William K. Funkhouser; Scott H. Randell; Philip S. Bernard; Charles M. Perou; D. Neil Hayes

Background Lung adenocarcinoma (LAD) has extreme genetic variation among patients, which is currently not well understood, limiting progress in therapy development and research. LAD intrinsic molecular subtypes are a validated stratification of naturally-occurring gene expression patterns and encompass different functional pathways and patient outcomes. Patients may have incurred different mutations and alterations that led to the different subtypes. We hypothesized that the LAD molecular subtypes co-occur with distinct mutations and alterations in patient tumors. Methodology/Principal Findings The LAD molecular subtypes (Bronchioid, Magnoid, and Squamoid) were tested for association with gene mutations and DNA copy number alterations using statistical methods and published cohorts (n = 504). A novel validation (n = 116) cohort was assayed and interrogated to confirm subtype-alteration associations. Gene mutation rates (EGFR, KRAS, STK11, TP53), chromosomal instability, regional copy number, and genomewide DNA methylation were significantly different among tumors of the molecular subtypes. Secondary analyses compared subtypes by integrated alterations and patient outcomes. Tumors having integrated alterations in the same gene associated with the subtypes, e.g. mutation, deletion and underexpression of STK11 with Magnoid, and mutation, amplification, and overexpression of EGFR with Bronchioid. The subtypes also associated with tumors having concurrent mutant genes, such as KRAS-STK11 with Magnoid. Patient overall survival, cisplatin plus vinorelbine therapy response and predicted gefitinib sensitivity were significantly different among the subtypes. Conclusions/ Significance The lung adenocarcinoma intrinsic molecular subtypes co-occur with grossly distinct genomic alterations and with patient therapy response. These results advance the understanding of lung adenocarcinoma etiology and nominate patient subgroups for future evaluation of treatment response.


Brain Pathology | 2009

Malignant gliomas with primitive neuroectodermal tumor-like components: A clinicopathologic and genetic study of 53 cases

Arie Perry; C. Ryan Miller; Meena Gujrati; Bernd W. Scheithauer; Sandro Casavilca Zambrano; Sarah C. Jost; Ravi Raghavan; Jiang Qian; Elizabeth J. Cochran; Jason T. Huse; Eric C. Holland; Peter C. Burger; Marc K. Rosenblum

Central nervous system neoplasms with combined features of malignant glioma and primitive neuroectodermal tumor (MG‐PNET) are rare, poorly characterized, and pose diagnostic as well as treatment dilemmas. We studied 53 MG‐PNETs in patients from 12 to 80 years of age (median = 54 years). The PNET‐like component consisted of sharply demarcated hypercellular nodules with evidence of neuronal differentiation. Anaplasia, as seen in medulloblastomas, was noted in 70%. Within the primitive element, N‐myc or c‐myc gene amplifications were seen in 43%. In contrast, glioma‐associated alterations involved both components, 10q loss (50%) being most common. Therapy included radiation (78%), temozolomide (63%) and platinum‐based chemotherapy (31%). Cerebrospinal fluid (CSF) dissemination developed in eight patients, with response to PNET‐like therapy occurring in at least three. At last follow‐up, 27 patients died, their median survival being 9.1 months. We conclude that the primitive component of the MG‐PNET: (i) arises within a pre‐existing MG, most often a secondary glioblastoma; (ii) may represent a metaplastic process or expansion of a tumor stem/progenitor cell clone; (iii) often shows histologic anaplasia and N‐myc (or c‐myc) amplification; (iv) has the capacity to seed the CSF; and (v) may respond to platinum‐based chemotherapy regimens.


Cancer | 2011

The prognostic contribution of clinical breast cancer subtype, age, and race among patients with breast cancer brain metastases

Carey K. Anders; Allison M. Deal; C. Ryan Miller; Carmen Khorram; Hong Meng; E. Burrows; Chad A. Livasy; Karen J. Fritchie; Matthew G. Ewend; Charles M. Perou; Lisa A. Carey

Brain metastases (BM) arising from triple‐negative breast cancer (TNBC) portend a poor prognosis. TNBC is more common in premenopausal and African‐American (AA) patients; both of these characteristics also confer a poor prognosis. In a single‐institution cohort study, the authors attempted to determine whether the inferior outcome noted with TNBC brain metastases is more reflective of a higher risk population or the subtype itself.


European Urology | 2014

ClearCode34: A Prognostic Risk Predictor for Localized Clear Cell Renal Cell Carcinoma

Samira A. Brooks; A. Rose Brannon; Joel S. Parker; Jennifer C. Fisher; Oishee Sen; Michael W. Kattan; A. Ari Hakimi; James J. Hsieh; Toni K. Choueiri; Pheroze Tamboli; Jodi K. Maranchie; Peter Hinds; C. Ryan Miller; Matthew E. Nielsen; W.Kimryn Rathmell

BACKGROUND Gene expression signatures have proven to be useful tools in many cancers to identify distinct subtypes of disease based on molecular features that drive pathogenesis, and to aid in predicting clinical outcomes. However, there are no current signatures for kidney cancer that are applicable in a clinical setting. OBJECTIVE To generate a signature biomarker for the clear cell renal cell carcinoma (ccRCC) good risk (ccA) and poor risk (ccB) subtype classification that could be readily applied to clinical samples to develop an integrated model for biologically defined risk stratification. DESIGN, SETTING, AND PARTICIPANTS A set of 72 ccRCC sample standards was used to develop a 34-gene classifier (ClearCode34) for assigning ccRCC tumors to subtypes. The classifier was applied to RNA-sequencing data from 380 nonmetastatic ccRCC samples from the Cancer Genome Atlas (TCGA), and to 157 formalin-fixed clinical samples collected at the University of North Carolina. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Kaplan-Meier analyses were performed on the individual cohorts to calculate recurrence-free survival (RFS), cancer-specific survival (CSS), and overall survival (OS). Training and test sets were randomly selected from the combined cohorts to assemble a risk prediction model for disease recurrence. RESULTS AND LIMITATIONS The subtypes were significantly associated with RFS (p<0.01), CSS (p<0.01), and OS (p<0.01). Hazard ratios for subtype classification were similar to those of stage and grade in association with recurrence risk, and remained significant in multivariate analyses. An integrated molecular/clinical model for RFS to assign patients to risk groups was able to accurately predict CSS above established, clinical risk-prediction algorithms. CONCLUSIONS The ClearCode34-based model provides prognostic stratification that improves upon established algorithms to assess risk for recurrence and death for nonmetastatic ccRCC patients. PATIENT SUMMARY We developed a 34-gene subtype predictor to classify clear cell renal cell carcinoma tumors according to ccA or ccB subtypes and built a subtype-inclusive model to analyze patient survival outcomes.


The American Journal of Gastroenterology | 2011

Tryptase staining of mast cells may differentiate eosinophilic esophagitis from gastroesophageal reflux disease

Evan S. Dellon; Xiaoxin Chen; C. Ryan Miller; Karen J. Fritchie; Tara C. Rubinas; John T. Woosley; Nicholas J. Shaheen

OBJECTIVES:Mast cells may contribute to the pathogenesis of eosinophilic esophagitis (EoE), but their role in diagnosis is unknown. Our aim was to determine whether tryptase staining of esophageal mast cells differentiates EoE from gastroesophageal reflux disease (GERD) and has utility for diagnosis of EoE.METHODS:We performed a case–control study comparing patients with EoE, defined by consensus guidelines, to GERD patients with eosinophils on esophageal biopsy. Immunohistochemistry was performed with mast cell tryptase. The density (mast cells/mm2) and intensity (0–4 scale) of mast cell staining was compared between groups after masking the diagnosis. Receiver operating characteristic (ROC) curves were constructed, and the area under the curve (AUC) was calculated to assess mast cell staining as both a stand-alone diagnostic test and an adjunctive assay with eosinophil counts.RESULTS:Fifty-four EoE (mean age 24 years; 69% male; mean 146 eosinophils per high-power field (eos/hpf)) and 55 GERD (mean age 34 years; 60% male; mean 20 eos/hpf) patients were analyzed. The maximum epithelial tryptase density was higher in EoE than in GERD (162±87 mast cells/mm2 vs. 67±54; P<0.001). Mast cells were diffusely distributed throughout the biopsy in more EoE than GERD patients (41 vs. 7%; P<0.001). Tryptase density and eosinophil count were only weakly correlated (R2=0.09; P=0.002). The AUC was 0.84 for tryptase staining alone, and 0.96 for the combination of mast cells and eosinophils.CONCLUSIONS:Patients with EoE have higher levels of tryptase-positive mast cells compared with GERD patients, improving the diagnostic value of biopsies beyond eosinophil counts alone. Mast cell tryptase may have utility as a diagnostic assay for EoE.

Collaboration


Dive into the C. Ryan Miller's collaboration.

Top Co-Authors

Avatar

Ryan E. Bash

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Ralf S. Schmid

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Robert S. McNeill

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Carey K. Anders

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Charles M. Perou

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Mark Vitucci

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Arie Perry

University of California

View shared research outputs
Top Co-Authors

Avatar

Leigh B. Thorne

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Marni B. Siegel

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Matthew G. Ewend

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge